You are on page 1of 15

Biomedicine & Pharmacotherapy 112 (2019) 108615

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Mechanistic insight into diabetic wounds: Pathogenesis, molecular targets T


and treatment strategies to pace wound healing
Satish Patel, Shikha Srivastava, Manju Rawat Singh, Deependra Singh

University Institute of Pharmacy, Pt. Ravishankar Shukla University, 492010, Raipur, C.G., India

ARTICLE INFO ABSTRACT

Keywords: Wound management in diabetic patient is of an extreme clinical and social concern. The delayed and impaired
Diabetes healing makes it more critical for research focus. The research on impaired healing process is proceeding hastily
Diabetic wounds evident by new therapeutic approaches other than conventional such as single growth factor, dual growth factor,
Wound healing skin substitutes, cytokine stimulators, cytokine inhibitors, matrix metalloproteinase inhibitors, gene and stem
Growth factor
cell therapy, extracellular matrix and angiogenesis stimulators. Although numerous studies are available that
Compromised wounds
Diabetic foot ulcer
support delayed wound healing in diabetes but detailed mechanistic insight including factors involved and their
role still needs to be revealed. This review mainly focuses on the molecular cascades of cytokines (with growth
factors) and erstwhile factors responsible for delayed wound healing, molecular targets and recent advancements
in complete healing and its cure. Present article briefed recent pioneering information on possible molecular
targets and treatment strategies including clinical trials to clinicians and researchers working in similar area.

1. Introduction alteration in normal physiological functions like deprived blood circu-


lation, obesity, diseases like diabetes and stressed environmental con-
Diabetes is a multifaceted metabolic disease that affects more than 340 ditions. Based on their healing potential wounds are indicated into two
million individuals and about 20% of them develop diabetic wounds forms i.e. chronic and acute one. Chronic wounds include tissue injuries
worldwide [1]. Leg or foot ulcers are most common wounds in diabetic which do not heal in an organized set of stages and takes more than 12
patients. Diabetic patients have declined ability to metabolize glucose weeks for healing [4,5]. Normally, healing process starts with hae-
resulting in hyperglycaemic conditions which further complicate the mostasis that checks the blood loss and invasion of microbes to
wound healing process. This can result in stalled chronic wounds. The wounded area. This phase is rapidly followed and overlapped by an
incidence of delayed healing process in diabetic patient is increasing inflammatory phase, in which pro-inflammatory cells neutrophils up-
globally due to lack of preventive and control measures. About 2.5%–15% regulate (initially) followed by macrophages which clean up debris and
of yearly world-wide health budgets are consumed on diabetes mellitus pathogens along with growth factors and other cytokines and cells.
and diabetic wounds stake a major part. WHO report speculate that dia- Proliferative phase overlaps inflammatory phase in which new tissue,
betes will be the 7th foremost reason for death in 2030. In 2014, 9% of new blood vessels (angiogenesis) and matrix construction is initiated to
adults had diabetes and was the reason for death of 1.5 million patients in fill the wounded area. The final remodelling phase then increases the
2012. More than 80% of diabetes deaths occur in low- and middle-income tensile strength of the extracellular matrix and reduces the blood supply
countries [1,2]. Approximately 50%–70% of all the limb amputations are to the damaged area [6–9].
because of diabetic wounds and it was reported that in every 30 s, one leg DWs1 are one of the major concerns which mainly includes leg
is amputated due to diabetic wounds in worldwide [3]. ulcer/diabetic ulcer. Diabetes delay healing process because it impairs
Wound healing is a multifaceted and dynamic process which results each phase of wound healing i.e. haemostasis, inflammation, pro-
in the restoration of anatomic integrity with analogous function. Prime liferation and remodelling phase, which has a long-term negative effect
requirement for wound management is rapid and complete healing on quality of life, morbidity and mortality (Fig. 1). DWs are char-
without spreading infection and sepsis. Acute wounds generally heal acterized by delayed acute wounds and chronic wounds unveiling im-
with ease without any issue. The major concern involves age related paired healing due to a postponed, incomplete, or uncoordinated


Corresponding author.
E-mail address: deependraiop@gmail.com (D. Singh).
1
Diabetic Wounds

https://doi.org/10.1016/j.biopha.2019.108615
Received 17 October 2018; Received in revised form 19 January 2019; Accepted 23 January 2019
0753-3322/ © 2019 Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

Fig. 1. Interruption of normal wound healing process in diabetes.

healing process. DWs exhibit a persistent inflammatory phase asso- β7 in both diabetic and non-diabetic conditions. Neutrophils along with
ciated with an impediment in the formation of mature granulation T and B cells are also a significant producer of TNF-α, IL-10 and other
tissue and reduction in wound tensile strength. This may be due to the cells, keratinocytes, fibroblasts, mast cells and endothelial cells. These
vascular damage resulting in ischemia [10,11]. Each wound is an alarm cells also contribute in production of VEGF, IGF-1 and TGF-β [113].
for health and needs instant care. Generally, wounds are of two types on Macrophages are pivotal contributor in healing. Hyperglycaemia and
the basis of origin- external and internal. External origin wounds are oxidative stress changes the epigenic code that results into change in
like cuts, injuries, burns and bruises. These external wounds may fre- macrophage polarization and its modulation. Dys-regulated macro-
quently go unnoticed by diabetic patient because of peripheral neuro- phage polarization is one of the main reasons in delayed wound healing
pathy. Internal origin wounds like skin ulcers and calluses cause de- [13,14]. Studies revealed that in diabetes, a complex mechanism in-
struction of skin and nearby tissues with chances of bacterial infection. volve at molecular level which is responsible for delayed wound
Existing standard approach employs series of medical treatments to healing. Activities like sustained production of pro-inflammatory cy-
clean and eradicate the infected tissue, and maintain moisture with tokines, impaired angiogenic response and microvascular complications
adequate blood supply [12]. Recent studies have engaged in under- [10], impaired macrophage and neutrophils function [15], impaired
standing the critical factors that influence healing process. Although a keratinocytes and fibroblast migration and proliferation and impaired
great deal remains to be learned, these studies may lead to therapeutics production of healing-associated factors like impaired growth factor
that will promote proper tissue repair and improve impaired wound production has been reported in diabetic animal models [16].
healing. Present review is an attempt to reveal the molecular events The phases of healing process in diabetic patients are also stalled by
responsible for delayed wound healing, molecular targets for complete other factors, including specific metabolic deficiencies, impaired phy-
healing and recent advancements in its management. siological responses like hypoxia due to glycation of haemoglobin and
alteration of red blood cells membrane and narrowing of blood vessels
[17]. Hypoxia involves decreased oxygen supply to wounds due to
2. Mechanistic insight narrowed blood vessels. Glycation of haemoglobin causes deficient
supply of nutrients and oxygen to tissue which further delay the healing
Diabetes delays the healing process leading to a non-healing wound process. Hypoxia/glucose deficiency and deformed proteins produce a
including various complications with associated psychiatric stress and stress response to cell by accretion of unfolded proteins within en-
depression. These complications consist of functional limitations, dif- doplasmic reticulum known as UPR8 . This UPR is activated just after
ficulty in walking, and infection like cellulitis, abscess, osteomyelitis, tissue or skin injury and linked to production of pro-inflammatory
gangrene, and septicaemia. Impairment of healing in diabetic patients mediators. DWs showed sustained induction of UPR along with aug-
is familiar but the link between patho-physiology and impaired wound mented expression of the pro-inflammatory chemokine as compared to
healing in diabetes is still an unknown etiology. The healing process normal wounds [18].
necessitates collaboration between inflammatory cells and biochemical Local ischemia due to microvascular complications in diabetes
mediators stimulated by various factors. However, alteration of the considerably delays wound healing. miRNAs9 are a class of noncoding
cellular and biochemical factors and activities have been implicated in RNAs of 19–24 nucleotides in length that link in numerous physiolo-
the failure of wound healing in diabetics. gical processes and play an important role in these complications. Al-
Cells involved in wound healing process are neutrophils, monocyte, tered levels of miRNAs have been reported in various diseases and
macrophages, keratinocytes, fibroblasts, T cells, B cells, mast cell and impaired wound healing [19]. One of the miRNAs, MiR-210 is induced
endothelial cells. These cells are actively involved in the production and in hypoxic situations and targets E2f3 that reduce keratinocyte pro-
regulation of various cytokines and growth factors. Monocyte, which liferation in wound healing [20]. MiR-200b decreases angiogenesis by
later transform into macrophages is the foremost producer of pro-infl- steering globin transcription factor 2 and VEGFR210 [21]. Similarly
ammatory cytokines IL-1β2, TNF-α3, IL-64 and VEGF5, IGF-16 and TGF- various miRNAs like miR-130a, miR-21, miR-146a, miR-198 and miR-

2
Interleukin-1β
3 7
Tumour necrosis factor-α Transforming growth factor-β
4 8
Interleukin-6 unfolded protein response
5 9
Vascular endothelial growth factor Micro RNA
6 10
Insulin-like growth factor-1 Vascular endothelial growth factor receptor

2
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

26a involve in diabetic wounds that affect epithelization, delay in- Abnormal glycation of nerve cell proteins and the unfortunate activa-
flammation, fibroblast migration, keratinocyte migration, re- tion of protein kinase C due to hyperglycaemia and oxidative stress lead
epithelialization and angiogenesis [22,23] to nerve dysfunction and ischemia [38]. Lack of protective sensation in
Along with these, there are some physiologic factors like increased diabetic wounds leads to unnoticed progression of wounds to worse.
serum matrix metalloproteinase-9 [24], impaired collagen accumula- Recently Huang et al., reported that alteration in the Akt/mTOR
tion and variation in the ratio of collagen types, dysregulation in the pathway results in impaired wound healing in diabetes induced rat
neuropeptide expression in the skin along with a suppressed infl- model [39]. Similarly, Lima et al reported, Insulin induces activation of
ammatory response [25], deficiency of thrombin-activatable fi- insulin signalling pathways i.e IR/SHC/ERK and IR/IRS/PI3K/AKT
brinolysis inhibitor [26], AGE11 modification of PDGF [27], decreased pathways in wound healing. By inducing these pathways it increases
number of epidermal nerves, epidermal barrier function [10] and VEGF and SDF1a tissue expression, increased eNOS phosphorylation,
misbalance between the accumulation of ECM components and their angiogenesis and improved healing in diabetes [40]. Studies revealed
remodelling by matrix metalloproteinase [17,28] are responsible for the role of matricellular proteins in wound healing which linked with
slow healing process in diabetic patients (Fig. 2). the proteins of the ECM and connecting them to cell surface receptors.
One of the matricellular proteins, AL-420 facilitates keratinocyte mi-
2.1. Roles of chemokines, free radicals and oxidative stress gration, reepithelialization and angiogenesis. AL-4 can connect with
integrin β1, and activate the SRC, ERK, and AKT signaling cascades and
In last decades, noteworthy evidences have been generated to sup- start JAK1/STAT3 activation. Activated STAT3 can induce the up-reg-
port a number of mechanisms as shown in Fig. 3 which influence dia- ulation of iNOS21 expression and increase NO production from the
betic wounds including polyol pathway, hexosamine pathway, dia- keratinocytes in the wound tissue and promote angiogenesis. Normally
cylglycerol pathway, nitric oxide blocking, PKC (protein kinase C) AL-4 expression has been low in normal skin and is significantly ele-
pathways, formation of AGEs i.e. maillard reaction and intraglomerular vated upon injury. But in case of diabetic patient, AL-4 expression re-
hypertension induced by glomerular hyperfiltration which lead to mains low, that delayed the healing process by affecting angiogenesis
neuropathy. These mechanisms are stimulated by mitochondrial over- and re-epithelialization [41].
production of reactive oxygen species [29] and oxidative stress. In
diabetes, high oxidative stress plays a major role in complications and
2.2. Role of immune system
impaired healing process. One of the transcription factor NRF212 con-
trols the adaptive response to oxidative stresses as well as decreased
Suitable co-ordination of innate immune system has an important
apoptosis, promote cell migration, proliferation, and cell differentiation
role in wound healing. TLRs22 are important source for the initiation of
[30]. High glucose and oxidative stress activates NRF2 to control and
the innate immune and inflammation response. Down regulation of
repair the impairment. Long et al had shown that inducing NRF2 ac-
TLRs-2 in injured tissue impairs or weakens the immune system and
tivation reduced diabetes-induced oxidative stress levels, regulates the
inflammation response [42–44] in diabetic patient which causes re-
expression MMP-9, TGF-β, migration and proliferation-related genes
duced chemotactic effect that delays the recruitment of various in-
via direct or indirect mechanisms [31].
flammatory cells. Diabetic patients are highly susceptible to infection
ATF-313 is a stress-inducible gene, and its expression is induced B-
caused by delayed wound healing and immuno-suppression [42]. Bac-
cell dysregulation and diabetic complications [32]. The irregular pro-
terial connections on the wound are important in the etiology of dia-
inflammatory response activates ATF-3 and iNOS and induces oxidative
betic wounds and it forms biofilms. These biofilms provide a safeguard
stress, which might be responsible for the prolonged healing processes.
to microbes from antimicrobial agents and immune system and inter-
Badr et al revealed that irrationally up-regulated expression of ATF-3
rupt the healing process. It is the most common reason of lower limb
and iNOS tailed by an increase in free radical levels and rise in caspase-
amputation in diabetic wounds [45].
3,-8, and -9 activityare responsible for impaired cellular differentiation
Inflammatory cells like neutrophils, monocyte, T cells, B cells and
and remodelling phase in healing process [33].
mast cell play a chief role in the immunity. Dysegulation of these cells
Numerous proinflammatory cytokines plays a major role in leuko-
may be decisive in the inhibition of diabetic host immunity. Elevated
cyte accumulation (monocytes /macrophages/ neutrophils/ immature
levels of pro-inflammatory cytokines such as IL-6 and TNF-α in diabetes
dendritic cells) like MIP1α14, MIP-215 and KC16 and human β-defensin
causes interrupted inflammatory cascade, hyperinflammation and in-
(HβD 1, 2, 3) as antibacterial in healing process. CX3CL117 is stated as a
sulin resistance. Elevated level of TNF-α may be due to accumulation of
soluble chemokine and membrane-bound form on the surface of cells
effector T cells. Maura et al found lower naive T-cell number and a
promoting macrophage and fibroblast accumulation [34–36]. Badr
poorer T cell receptor (TCR-Vβ) range in diabetic wound patients that
et al. revealed that abnormal expression of MIP1 α, MIP-2, CX3CL1,
lead to the accumulation of effector T cells [46].
reduce level of HβD 1, 2 and 3, irregular activation of STAT318 and
Immunity is also influenced by high level of AGEs inducing pro-
decline in the activation of AKT/PKB (serine/threonine protein kinase
duction of various cytokines such as IL-6 and TNF-α. AGEs also obstruct
B) and NF-κB19 . All these collectively contribute tointerrupt healing in
the collagen production, induces apoptosis, excessive immune-re-
diabetic wounds [34,37].
sponses and negative regulations of the cell physiology leading to im-
Diabetic peripheral neuropathy lead to sensory, motor and auto-
paired healing [47]. Mast cells are potent releaser of angiogenic factors
nomic dysfunction, each of which contributes to delayed wound
like FGF, VEGF, and TGF-β1 [48]. Various studies support the role of
healing. In sensory neuropathy, sensation to pain either is lost or absent
mast cells in wound healing as they act together with macrophages,
creating most important threat for the growth of diabetic wounds.
endothelial cells, and fibroblasts. It plays an important role in matrix
restructuring and disturbs the balance between pro-angiogenic factors
11
Advanced glycation end-products and anti-angiogenic factors in wound tissues [49]. Bevan et al revealed
12
Nuclear factor-E2-related factor 2 that in genetically diabetic mice, there was delayed vascular regression
13
Activating transcription factor-3 due to decrease in the number of mast cells and their dysfunction [50].
14
Macrophage inflammatory proteins 1α Nishikori et al demonstrated the role of mast cell in diabetic wound.
15
Macrophage inflammatory proteins 2
16
Keratinocyte derived chemokines
17 20
Fractalkine Angiopoietin-like 4
18 21
Signal transducer and activator of transcription 3 Inducible nitric oxide synthase
19 22
nuclear factor-κB Toll like receptors

3
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

Fig. 2. Factors responsible for Diabetic Wounds.

Fig. 3. Major pathways responsible for decreased wound healing in diabetes [4].

They concluded impaired proliferative phase in healing process in assist endothelial cell migration, stimulate pro-collagen synthesis and
diabetes due to delayed increment of mast cells. The dysfunction of protein homeostasis [43,53]. In diabetes, level of HSPs (HSP90, HSP70,
mast cells affects the angiogenesis in proliferative phase and vascular HSP47 and HSP27) decreases along with their downstream molecules
regression in the remodelling phase [48]. Tellechea et al, revealed in his TLR4 and p38-MAPK [54] that might be responsible for impaired
experiment that mast cell degranulation was enhanced in the skin of healing process.
humans and mice with diabetes that led to impaired healing process.
Mast cell degranulation inhibitors like disodium cromoglycate, quer-
3. Role of Growth factors in impaired wound healing
cetin, and luteolin might be a potent drugs to improve wound healing in
diabetes [49]. The RNA expression of MIF (Macrophage Migration In-
Normal wound healing cascade is well coordinated and synchro-
hibitory Factor) genes deceases in diabeties, which is an important
nised by growth factors, different MMPs (Matrix metalloproteinase),
molecule in pro-inflammatory innate immune reactions. Studies re-
cytokines, inflammatory cells, keratinocytes, fibroblasts and en-
ported that decreased level of MIF in diabetic wound might be re-
dothelial cells. Growth factors are biologically active polypeptides
sponsible for impaired production of endothelial progenitor cells and
which are involved in all phase of healing process [55]. They promote
healing process [51,52].
the early inflammatory phase during the granulation phase of tissue
Heat shock proteins (HSPs) promotes wound healing by recruiting
formation. Compromised wounds frequently show defects in the type
dermal fibroblasts, stimulate cell proliferation, differentiation of kera-
and amount of growth factor due to change of their expression, de-
tinocytes, decreases oxidative stress, alleviating actin microfilaments,
creased production, decreased release, trapping and excessive

4
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

degradation [56]. Balance between matrix formation and matrix de- wound healing process [76]. TGF-β recruits and promotes stimulation
gradation characterizes ECM synthesis with optimum healing. Factors of inflammatory cells neutrophils, macrophages, lymphocytes, kerati-
regulating ECM formation like VEGF [57], IGF-I, IGF-II [58], TGF-β23 nocytes, fibroblasts and production of growth factors. These accelerate
[59], KGF24 [60], PDGF25 [61], EGF26 [62], FGF27 [63,64], TNF- α and vascularisation, angiogenesis, and formation of ECM and hindered de-
IL-6 are noticeably decreased in diabetic patients. Growth factors play a gradation of ECM [77]. The reduced concentration of TGF-β has been
critical role in initiating and sustaining the different phases of wound reported in wound healing in diabetic case [59]. Various studies de-
healing (Fig. 4). Any alteration i.e. down-regulation of growth factor monstrated that MMP-encoding genes have a TGF-β1-dependent in-
receptors and rapid degradation of growth factor leads to delayed hibitory element in the promoter region, which down-regulate the
wound healing in diabetics. gene’s expression. Decreased levels of TGF-β and increased expression
Platelet releases the PDGF that is a key serum mitogen and induces of MMPs causes excessive degradation of the growth factors [78]. Along
fibroblast proliferation, matrix production, and maturation of con- with MMP-encoding genes, Transcription factors like Smad-2, Smad-3
nective tissue [65]. PDGF is synthesized continuously in the wound and Smad-4 also activate and repress TGF-β target genes. TGF-β1 ac-
milieu by macrophages which are major cell in the late inflammatory tivates the Smad-2 and 3 for production of collagen [79]. Decrease in
phase. PDGF works as a chemo-attractant for fibroblasts and in- level of TGF-β1 lead to increased recruitment of activated inflammatory
flammatory cells. It facilitates synthesis of glycosaminoglycans, pro- cells causing delayed inflammatory phase to proliferation phase of
teoglycans and collagen. It acts as a key mediator in the migration and healing process in DWs. High level of TGF-β3 was thought to be the
proliferation of fibroblasts, production of granulation tissue proteins reason for decreased level of TGF-β1 in diabetics [80] which lead to
and provisional ECM and angiogenesis during the healing process [66]. increased macrophage activity and decreased collagen synthesis. In
PDGF and its receptors expression is decreased in diabetic wounds, diabetics, high glucose level augmented macrophage activity producing
signifying its role in healing process [67]. Various clinical studies using more reactive oxygen species leading to prolonged inflammatory phase
PDGF have shown enhanced healing time [68]. [81]. Decreased level and expression of these growth factors lead to
bFGF28 posses stimulatory effect on the growth and differentiated poor and prolonged wound healing process in diabetes.
function of fibroblasts and on the proliferation of vascular smooth
muscle cells, endothelial cells, extracellular matrix metabolism, growth,
and movement of mesodermally derived cells. It increases the rate and 4. Role of MMPs in impaired wound healing
degree of granulation tissue formation and stimulates healing process
[69]. Matrix metalloproteinase is a family of 26 zin. dependent en-
VEGF is one of the most potent known angiogenic cytokines in the dopeptidases that play crucial role in initial wound debridement, as
skin, and the amount of VEGF present in a wound can notably impact well as in the phases of angiogenesis, epithelialization, and remodelling
healing and supports rate-limiting steps of vasculogenesis and angio- of extra cellular matrix [82]. Every matrix proteins like collagens,
genesis. It mainly involves in deterioration of the extracellular matrix of basement membrane collagens, proteoglycans, elastin, fibronectin are
existing vessels by proteases and causes migration and proliferation of digested by MMPs. The gelatinase (MMP-2 and MMP-9) are two pro-
capillary endothelial cells [70]. VEGF increases capillary density in teinases primarily responsible for breaking down of type IV collagen
diabetic wounds and improves the blood perfusion and metabolism in from the basic matrix. These are present as inactive zymogens that need
injured tissue. Restoration of blood flow to injured tissues facilitates removal of the pro domain for their activation. Activity of MMPs is
oxygen and nutrients supply to support the growth and task of re- regulated by complexation with TIMPs29, which block contact to the
parative cells which promotes wound healing. It is the main regulating active site. Balance between MMPs and TIMPs is required for a proper
factor in the revascularization and permeability of the wound site, and wound healing as supported by number of studies [83]. MMPs are in-
participates in the formation of the granulation tissue. Roles of VEGF be volved in various stages of wound healing like cell migration by pro-
governed by activation of its receptors, first receptor VEGF receptor-1 teolysis of the ECM, in re-epithelialization by degradation of junctional
activation leads to inflammation while activation of VEGF receptor-2 proteins, in leukocyte invasion by creating a chemotactic gradient, in
lead to angiogenesis [71]. The relatively low level of VEGF in local inflammation by processing of multiple cytokine either by inhibition or
wound is due to diabetes responsible for impaired wound healing. Re- by activation [84]. In these enzymes MMP-1 is crucial for wound re-
search studies found that abnormal patterns of VEGF receptors, de- epithelialization, MMP-2 is significant during angiogenesis and pro-
creased VEGF mRNA levels, increased VEGFR-1 level and decreased longed matrix remodelling and MMP-3 is vital for normal wound con-
VEGFR-2 level is main reason for non-healing status of wounds [72]. traction and in remodelling the basement membrane. While role of
Platelet releases EGF augmenting the epidermal cell, cell motility, MMP-9 during healing is indistinct; it may be involved in separating
cellular migration, mesenchymal regeneration, angiogenesis and cell keratinocyte from the basement membrane before migration and be
proliferation after binding to the EGF receptor [73]. IGF-1 and IGF-2 used to assist matrix degradation by neutrophils and macrophages for
are peptide that forms a complex Insulin-like growth factor (IGF). IGF-1 the period of elimination of necrotic or damaged tissue [85]. Studies
contributes in wound healing by participating in cell granulation and revealed that high levels of metalloproteases are a feature of diabetic
re-epithelisation, stimulating chemotaxis of endothelial cells and pro- wounds, and the MMP levels in chronic wound fluid are almost 60 times
liferation of keratinocytes and fibroblasts. However, in diabetes pa- higher than acute wounds. This increased protease activity supports
tients, expression of IGF-1 is decreased which may be the reason for cell tissue destruction and inhibits normal repair processes [86]. One of the
granulation imperfection [74,75]. Its affinities are modulated by the pH possible reasons behind this is high glucose concentrations that directly
of the wound environment [16]. alter the level and expression of MMPs, decrease the expression of
The decreased levels of IGF-1 and TGF-β at the wound tissue were TIMPs via effects of persistently high levels of pro-inflammatory and
reported in both diabetic animals and human responsible for delayed pro-fibrotic cytokines due to increased activation and invasion of in-
flammatory cells, and indirectly affect MMPs by formation of advance
glycation products [68]. These consequently abolish growth factors,
23 receptors, and matrix proteins crucial for wound healing.
Transforming growth factor β
24
Keratinocyte growth factor
25
Platelet derived growth factor
26
Epidermal growth factor
27
Fibroblast growth factor
28 29
Basic fibroblast growth factor Tissue inhibitors of matrix metalloproteinase

5
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

Fig. 4. Role of major Growth Factors on different cells and process involved in wound healing.

5. Molecular targets performed a clinical trial using photodynamic therapy and result shown
that rate of amputation in the photodynamic therapy group was 0.029
Increased understanding of diabetes and its role in impaired wound times the rate in the control group [87]. Park et al., 2018 performed a
healing at molecular level have broadened the wound research over phase III multicenter, double-blind, randomized, placebo controlled
recent years. These studies revealed that impaired wound healing in trial to evaluate the efficacy and safety of a novel spray-applied growth
diabetics is due to unusual cellular expression of all participating cells factor therapy containing recombinant human epidermal growth factor
and dys-regulation in the expression of cytokines, growth factors and (rhEGF) for the treatment of diabetic wounds. This group concluded
various other molecular factors required for coordinating the normal that patients in the rhEGF treated group notably completed healing as
healing process. As a result, chronic non-healing wounds are unable to compared to placebo group (73.2% vs 50.6%, respectively; p = 0.001).
step forward in synchrony and are checked mainly in the inflammatory Also, healing velocity was found to be faster in the rhEGF treated group
phase. Numerous molecular factors/targets for management of diabetic (p = 0.029) in spite of HbA1c levels. The rhEGF treated group had a
wounds have been identified over the years. These management ap- shorter median time to 50% ulcer size reduction and shorter time to
proaches are categorized on the basis of molecular targets which di- complete ulcer healing as compared to placebo group [54]. Asadi et al.,
rectly or indirectly modulates their activity. 2017 performed a randomized, single-blind, placebo-controlled trial
The targets, which directly interact, include various growth factors using low-intensity cathodal direct current. Results of this study sug-
(PDGF, TGF-α, EGF, VEGF, FGF, and KGF), autologous keratinocytes or gested that on applying electric stimulation to ischemic ulcers have
autologous fibroblasts and stem cells. The role and importance of these positive effects and promote healing by stimulate the release of HIF-1α
targets were discussed in previous sections. Various agents might in- and VEGF in the wound area [88]. Soleimani et al., 2017 performed a
directly affect the molecular targets by up-regulated or down-regulated randomized, double-blind, placebo-controlled trial using flaxseed oil
expression of growth factor, pro and anti inflammatory cytokines, omega-3 fatty acids. This group concluded that on supplementation of
MMP, nitrous oxide level, collagen synthesis /degradation and factors omega-3 fatty acids for 12 weeks among diabetic wound patients had a
promoting angiogenesis depending on the specific target. These wound favorable effects on parameters of ulcer size, indicators of insulin me-
management approaches are classified on the basis of types of ther- tabolism, plasma TAC, serum hs-CRP, and GSH levels [89]. Similar
apeutic agents used as drugs, growth factor, other approaches and stem various clinical trials have been performed in recent years to promote
cells and highlights of available clinical status for treatment of diabetic healing in diabetic wound patients was described in Table 4.
wounds are shown in Table 1. Table 1 also shown the molecular targets
directly or indirectly involve in therapeutic approaches by formations
or system or drug. 7. Future therapeutic strategies

5.1. Natural product based treatment Diabetic wounds consequences from numerous risk factors in-
cluding peripheral neuropathy, peripheral artery disease and foot ail-
Traditionally natural source obtained from different sources have ments. In spite of the development in technologies such as bioengi-
been of prime importance to heal wound like turmeric, castor leaves, neered skin cells and the prevalent application of standard care in
neem bark, rosemary, ginseng and many more. It has been reported that treating diabetic wounds, it has been reported that the occurrence of
70% of commercialized products contains plant based active in- wound healing has remained at less than 50%. The research on diabetic
gredients, rest 20% are of mineral base and 10% are of animal based wounds is proceeding hastily evident by new therapeutic approaches
and more than 13,000 have been worked out particular to accelerate other than conventional such as single growth factor, dual growth
the process of wound healing. Plant actives which are involved in factor, skin substitutes, cytokine stimulators, cytokine inhibitors, matrix
healing process include glycosides, steroids, saponins, resins, mucilage metalloproteinase inhibitors, gene and stem cell therapy, extracellular
and flavonoids. Table 2 enlists most of the medicinally active plants matrix and angiogenesis stimulators. Future treatment approaches are
which were demonstrated for their wound healing activity in diabetes at present under analysis for the treatment of diabetic wounds includes
and Table 3 depict the available marketed products used in manage- recombinant growth factors, platelet-rich plasma, Sphingosine 1-phos-
ment of diabetic wounds. phate, Substance P, stem cell therapy, MMP inhibitors, shock-wave
therapy, laser therapy and natural product based treatment. These ap-
6. Clinical trials proaches are the crucial need for a more realistic, secure and efficient
therapy for diabetic wounds (Tables 1 and 2). The majority of these
Various clinical trials were on going to uncover a novel treatment approaches are currently under investigation and their use has mostly
approach for this worldwide health ailment. Tardivoe et al., 2014 been restricted to clinical trials.

6
Table 1
Therapeutic strategies explored management of wounds in diabetics.
Therapeutic agents Delivery System and Route Molecular Target Outcome and Mechanism Target tissue/ animal Reference
S. Patel, et al.

model

Growth factors
PDGF/TGF-α Topically/ Gel PDGF and TGF-α Promoting healing by increasing keratinocyte and fibroblast Genetically diabetic [58]
mitogen mouse
basic fibroblast growth factor PELA Nanofibres/ Topically Fibroblast Enhanced proliferation, migration, fibroblast adhesion, re- Diabetic rats [90]
epithelization, angiogenesis, ECM restoration and remodelling
Human epidermal growth factor Cream EGF Reduces the healing time Diabetic foot ulcers [91]
Recombinant epidermal growth factor PLGA Microspheres rhEGF Increased healing by inducing fibroblasts proliferation and Diabetic ulcers [92]
enhancing proliferating cell nuclear antigen in the epidermis
PDGF Gel PDGF Increased angiogenesis, cell proliferation and epithelialization Diabetic rats [67]
basic fibroblast growth factor collagen/gelatin sponge ——— Improve healing Chronic skin ulcers [93]
Platelet Hyaluronic acid Gel PGDF, TGF-α, VEGF Enhance granulation tissue formation, re-epithelization and Cutaneous chronic [56]
reduce pain wounds
Recombinant PDGF Gel PDGF Increases reepithelialization, granulation tissue thickness, Diabetic rats [94]
capillary density and improved healing by increased c-fos protein
expression and ERK phosphorylation
Fusion protein (CBD-VEGF) Collagen Domain VEGF Increases vascularisation and maintain VEGF activity Diabetic rat model [95]
Recombinant human acidic fibroblast Chitosan crosslinked collagen sponge FGF Increases angiogenesis, collagen generation and dermal cell Diabetic rats [96]
growth factor proliferation
Recombinant epidermal growth factor Poly-epsilon-caprolactone and poly- EGF Promotes proliferation and expression of keratinocytes Diabetic ulcers [97]
ethyleneglycol Nanofibres
Arginine and Epidermal growth factor Hyaluronic acid sponge EGF Decreased wound size by increasing the epithelization [98]
bFGF Collagen-gelatin sponge FGF Accelerate ECM formation and angiogenesis Diabetic mouse model [99]
Keratinocytes growth factor Chimeric Nanoparticles KGF Increased healing by enhancing reepithelialization and Chronic wounds [100]
granulation tissue formation

7
plasmid bFGF Poly(Ethylene Imine) electrospun Fibers FGF Increased healing by enhanced collagen synthesis, maturation Diabetic skin wound [101]
and reepithelialization
rhEGF Dextrin conjugated Topically ——— Accelerated wound closure, neo-dermal tissue formation, (Db/db) diabetic [73]
increased granulation tissue deposition and angiogenesis mouse
Dual growth factor Poly(lactic-co-glycolic acid) nanoparticles in VEGF, PDGF-BB Accelerated the healing process by increasing angiogenesis, re- ——— [102]
chitosan nanofiber system epithelialization and granulation tissue formation
PDGF Collagen Chitosan hydrogel/ Topically ——— Increased collagen biosynthesis and by reducing reactive oxygen Cutaneous wound [103]
species healing
PDGF-BB Carboxymethyl cellulose Hydrogel/ Topically ——— Accelerated healing by enhanced granulation tissue formation Genetically diabetic [104]
and angiogenesis mice
bFGF Chitosan film/ Topically FGF Reduced wound area and promoted healing by increasing ECM Genetically diabetic [105]
formation mice
Recombinant human Epidermal Polyurethane foam/ Topically ——— Accelerated healing by increasing contraction rate, re- Diabetic wounds [106]
growth factor epithelialization and collagen deposition
VEGF Poly(lactic-co-glycolic acid) nanoparticle VEGF Receptor -2 Enhanced proliferation and migration of keratinocytes and Non-diabetic and [107]
upregulated the expression of VEGFR2 at mRNA level diabetic wounds
Synthetic drugs
Pravastatin Subcutaneous sponges eNOS expression Improve wound breaking strengths, hydroxyproline accumulation Diabetic wound [108]
by up-regulation of eNOS and NO expression
Azelnidipine Solution/Orally eNOS Accelerated healing by stimulating NO production and enhancing Skin Wound in [109]
histologic processes Diabetic Rats
AL–CS–PGA hydrogel Hydrogel Collagen Increases healing by collagen regeneration and epithelialization Diabetic rats [110]
Atorvastatin gel Carbopol Hydrogel ——— Increased healing with closure and epithelisation within 7 days Diabetic rats [111]
Erythropoietin Cream VEGF Decreased wound closure time, increased VEGF and Diabetic rats [112]
hydroxyproline and microvascular density
Pentoxifylline Cream MMPs and TIMP-1 Accelerates healing by decreasing MMPs expression and increased Diabetic rats [113]
TIMP-1 expression
(continued on next page)
Biomedicine & Pharmacotherapy 112 (2019) 108615
Table 1 (continued)

Therapeutic agents Delivery System and Route Molecular Target Outcome and Mechanism Target tissue/ animal Reference
S. Patel, et al.

model

Deferoxamine Intra-peritoneally HIF-1α, SDF-1α, VEGF Increased neovascularisation and healing by promoting Diabetic rats [114]
endothelial tube formation and cell proliferation through up-
regulation of HIF-1α
Topically ——— Increased neovascularisation and healing by decreasing mRNA Diabetic rats [115]
expression and protein levels of TNF-α, MMP-9 and IL-1β
Substance P I.V. endothelial cell adhesion molecules or Increases early leukocyte and macrophage density in healing and Diabetic murine [116]
IL-8 promoting cutaneous wound repair wounds
Topical application ——— Decreased levels of TNF-α, IL-1β and MMP-9 and increase IL-10 Diabetic rat [117]
levels and increased the expressions of VEGF, TGF-b, SDF-1α, HO-
1 and e-NOS.
Reverses the proinflammatory state in diabetic wound and Diabetic skin wounds [118]
modulate activation of macrophage and improve healing
Propranolol Solution Orally VEGF, TGF-β, IL-8, MMP-9 Reduces inflammatory cell and MMP-9 levels and increases cell Diabetic rats [119]
proliferation, mast cell number, collagen deposition, blood vessel
density, and nitric oxide level
Glucophage (metformin) nanofibrous collagen/PLGA membrane MMP-9 Faster healing by increased re-epithelialization and collagen I Diabetic wound [120]
synthesis by down-regulating MMP-9
Novel nano-insulin Silver nanoparticles Coated with Insulin IL-6, TNF-α, IL-10 Regulation of balance among IL-6, TNF-α and IL-10 at the wound Diabetic rats [121]
site to promote faster wound remodeling
GW501516 Polymer Microparticle/ peroxisome proliferator-activated Reduced the oxidative wound micro-environment to accelerate Diabetic wound [122]
receptor healing
MK0626 Dipeptidyl peptidase-4 inhibitor/ Oral HIF-1α / SDF-1 Significantly improved healing, angiogenesis, and endogenous Diabetic mice [123]
progenitor cell recruitment.
Poly (caprolactone) /gelatin Conducive Poly (caprolactone) /gelatin Epithelial-to-mesenchymal transition Improve diabetic wound healing by releasing Si ions and ano- Diabetic mice [124]
nanofibrous composite scaffold nanofibrous composite scaffold containing and endothelial mesenchymal fibrous structure

8
silicate-based bioceramic particles transformation pathway
Adenine Ointment AMP-activated protein Kinase, PPARδ Increasing the healing by increasing angiogenic related protein Diabetic mice [125]
PPARδ and decrease the AGE receptors.
Bee venom Subcutaneous injection Nrf2, Ang-1 and Tie-2 signaling Enhanced wound by rising collagen and BD-2 expression and Diabetic mice [126]
reinstate the levels of Ang-1 and Nrf2 and Tie-2 downstream
signaling
Curcumin Composite graft with Cytomodulin coupled —— Better and faster wound healing with regenerating a skin of Diabetic rats [127]
porous PLGA microparticles higher tensile strength.
Other approaches
Sphingosine 1-phosphate Subcutaneous EDGs Accelerates healing by increased vascular formation within the Diabetic mice [128]
granulation tissue
Adiponectin TGF-β Restrain proliferation and differentiation of keratinocytes, and Diabetic wound [129]
regulates the expression of TGF-β
Neurotensin 5-methyl pyrrolidinone Chitosan Dressing TNF-α, IL- 1β, collagen expression Reduced inflammatory status, improve healing, re- Diabetic wound [130,131]
epithelialization by decreasing inflammatory infiltrate and
increasing fibroblast migration and collagen deposition
Collagen dressings TNF-α, IL-1β Reduced inflammatory cytokine TNF-α, IL-1β and MMPs, Diabetic mice
increased fibroblasts migration and collagen expression and
deposition
Broad-spectrum MMP inhibitor Sepharose resin, Topically MMP and TIMP Accelerate healing, re-epithelialization and inhibit MMP-9 Diabetic wound [28]
activity
Hyaluronic acid Solution, I.P. TGF-β Improved healing by increasing skin remodelling proteins, TGF-β Genetically diabetic [132]
and transglutaminase-II mice
HoxD3 plasmid DNA Methylcellulose Film HoxD3, Col1A1 and β3-integrin Significant acceleration of wound closure by increasing mRNA Diabetic mice [133]
expression of HoxD3, Col1A1 and β3-integrin leading to
enhanced angiogenesis and collagen deposition
Human Amniotic fluid Proteome ——— ——— Accelerates healing by activating mitosis and angiogenesis Diabetes-impaired [134]
wound
(continued on next page)
Biomedicine & Pharmacotherapy 112 (2019) 108615
Table 1 (continued)

Therapeutic agents Delivery System and Route Molecular Target Outcome and Mechanism Target tissue/ animal Reference
S. Patel, et al.

model

Angiopoietin-like 4 Topical Nitric Oxide Increases diabetes induce wound healing via increasing nitric Diabetic mice and [41]
oxide production accelerated reepithelialization and improving human skin tissue
angiogenesis
Stem cells
Adipose tissue-derived Mesenchymal Intra-dermally VEGF, HGF Enhances healing by anti-inflammatory and anti-apoptotic effects Diabetic rats [135]
Stem Cells
Autologous Keratinocytes or Cell suspension Fibroblast, Keratinocyte Accelerate healing by enhancing re-epithelialization rate Diabetic porcine [136]
Autologous Fibroblasts wound
Allogenous skin fibroblasts Cell suspension Fibroblast Promote healing by increasing re-epithelialization, fibroblasts Diabetic sheep [137]
and angiogenesis
BM derived Mesenchymal Stem Cells Subcutaneously Cell suspension ——— Stimulate healing by increasing production of cytokines and/or Murine and human [138]
by stimulation of endogenous resident cells cutaneous wounds
EGF, VEGF, Prolyl4-hydroxylase, Ki-67 Enhances healing by reduction in topical pro-inflammatory Diabetic rats [139]
expression reaction and increases VEGF
Mesenchymal Stem cells ————— ————— Promotes healing by enhancing angiogenesis Diabetic wound [140]
Electrospun Collagen Scaffold Fibroblast Enhancing healing by promoting fibroblast migration and Diabetic mice [141]
proliferation
Topical embryonic stem cells Topical injection EGF, VEGF Accelerated healing by stimulating epidermal regeneration, Diabetic rats [142]
granulation tissue formation and angiogenesis via increasing EGF,
VEGF and fibronectin
Amniotic Mesenchymal Stem Cells Injection angiogenic factors Promotes healing by up-regulation of angiogenic factors, IGF-1, Diabetic NOD/SCID [143]
EGF and IL-8 and enhanced engraftment/ differentiation mice
capabilities
Placenta mesenchymal stem cell Intradermal injection Pro-angiogenic molecules Accelerates healing by stimulating vascular regeneration Diabetic Goto- [144]
Kakizaki rats

9
BM-derived SPCs Topical ————— Improves healing via indirect mechanisms that enhanced Diabetic mice [145]
angiogenesis
Adipose tissue-derived stromal cells Atelocollagen matrix ————— Accelerating healing by increasing granulation tissue formation, Db/db mice [146]
epithelium, and capillaries
Dermal sheath derived Mesenchymal ——— IL-6, IL -8 and growth-related oncogene Enhanced keratinocytes, fibroblasts proliferation and endothelial Diabetic wound [147]
Stromal cells cells in vitro and decreases healing time
Umbilical cord blood-derived 3D fibrin gel with CD34+-derived endothelial IL-17, IL-10, ERK1/2 patway Reduces the inflammatory reaction and enhances Diabetic mice [148]
hematopoietic stem cells cells neovascularization
(CD34+cells)
Adipose-derived stem cells Injectable hydrogel system CD11b, TNF α, IL-1 Accelerated healing by inhibiting inflammation and promoting Humanized excisional [149]
angiogenesis and re-epithelialization. wound model
Adipose-derived stem cells, Injection —— Increase in the % of wound closure rates in cell-based treatments. Diabetic swine [150]
Endothelial-differentiated stem
cells
Human umbilical cord blood–derived Transplantation Collagen and (TGF)-β Improved the healing by increasing collagen synthesis and Diabetic Mice [151]
mesenchymal stromal cells angiogenesis.
Biomedicine & Pharmacotherapy 112 (2019) 108615
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

Table 2
Natural product based treatment for diabetic wounds.
Natural products Outcome with possible mechanism Animal model Ref.

Rosmarinus officinalis L. Reduced inflammation, debridement, increased contraction, epidermal regeneration and alloxan induced-diabetic mice [152]
Family: Lamiaceae organization
Curculigo orchioides G. Improve healing by increasing superoxide dismutase, nitric oxide and decreased lipid Streptozotocin induced diabetic [153]
Family: Hypoxidaceae peroxidation mice
Melilotus officinalis (L.) Pall. Induces micro-vascularisation and Anti-inflammatory activity Diabetic patients [154]
Family: Fabaceae
Rehmanniae Libosch. ex Fisch. & C.A. Improved healing by enhancing tissue regeneration, angiogenesis and inflammation Streptozotocin-induced diabetic [155]
Mey. control rat
Family: Orobanchaceae
Curcumin Curcuma longa L. Increased wound closure rate by reduced inflammatory induction and antioxidant activity Streptozotocin induced diabetic [156]
mice model
Family: Zingiberaceae Improved healing by increasing granulation tissue, fibroblasts proliferation and collagen Streptozotocin induced diabetic [157]
deposition rats
Enhanced healing by accelerated neovasculogenesis, increased expressions of VEGF, TGF- Streptozotocin induced diabetic [158]
β, hypoxia-inducible growth factor-1α, stromal cell-derived growth factor-1α, and heme rats
oxygenase-1
Martynia annua L. Enhanced healing by free-radical scavenging activity of the flavonoids and luteolin Streptozotocin induced diabetic [159]
Family: Martyniaceae rats
Genistein Improved healing and angiogenesis by suppression of FoxO1, iNOS activity and oxidative STZ-induced type 1 diabetic mice [160]
Genista tinctoria stress
L.
Family: Fabaceae
Lithospermun erythrorhison Siebold & Decreased vascular permeability, increased granulation tissue formation Diabetic mice [161]
Zucc.
Family: Boraginaceae
Astragalus membranaceus (Fisch.) Increased healing and post-ischemic neovascularization by augmenting blood vessel STZ induced rat with hindlimb [162]
Family: Fabaceae, density, VEGF and eNOS expression, and attenuate oxidative stress ischemia model
Rehmannia glutinosa (Gaertn.) Steud.
Family: Orobanchaceae
Astragalus membranaceus (Fisch.) Enhanced healing by increased tissue regeneration, promoting angiogenesis and Streptozotocin-induced diabetic [163]
Family: Fabaceae, inhibiting inflammation rat
Rehmanniae Libosch. ex Fisch. & C.A.
Mey.
Family: Orobanchaceae
Annona squamosa L. Enhanced epithelialization rate, cellular proliferation and collagen synthesis Streptozotocin-induced diabetic [164]
Family: Annonaceae rat
Nicotine Accelerated healing and angiogenesis Streptozotocin induced diabetic [165]
Nicotiana tabacum L. mice
Family: Solanaceae
Allium sativum L. Improve healing in diabetes Alloxan induced diabetic rats [166]
Family: Amaryllidaceae
Naringin Enhanced healing by inducing angiogenesis and down-regulate the expression of TNF-α, Streptozotocin-induced diabetic [167]
IL-1β and IL-6 and upregulate the expression of IFG-1, VEGF and TGF-b rat
Aloe vera (L.) Burm.f. Accelerated healing Streptozotocin-induced diabetic [168]
Family: Asphodelaceae rat
Sparassis crispa (Wulfen) Fr. Improve healing by promoting migration of macrophages, fibroblasts, and synthesis of streptozotocin induced diabetic [169]
Family: Sparassidaceae type I collagen. rats
Honey Promotes epithelisation Human patients [170]
Apis mellifera L.
Family: Apidae
Honey with hydroalginate Improve wound healing Human patients [171]
Astragulus polysaccharide-loaded Accelerated healing by restoration of microcirculation and promoted angiogenesis Streptozotocin-induced diabetic rat [172]
fibrous mats
Angelica sinensis (Oliv.) Diels, Improve healing by reduced neutrophil infiltration and macrophage accumulation, Streptozotocin-induced diabetic [173]
Family: Apiaceae enhanced angiogenesis, and increased collagen deposition rat
Astragalus membranaceus (Fisch.)
Family: Fabaceae,
Angelica dahurica Fisch.ex Hoffm.,
Family: Apiaceae and Gleditsia sinensis
Lam.
Family: Fabaceae
Bee venom Enhanced wound closure by increasing collagen production and reinstating the levels of Type I diabetic mouse model [33]
inflammatory cytokines by acting on ATF-3 and iNOS
Camel milk Peptide Restore the normal redox status and activate the inflammatory cascade and stimulates Streptozotocin-induced diabetic [52]
healing rat
Whey Protein Improves healing by increased glutathione synthesis and cellular antioxidant defence Streptozotocin-induced diabetic [37]
mice
Improved healing by Up-regulation of Hsp72 and keratin16 Streptozotocin-induced diabetic [174]
rat
Enhanced collagen deposition, restored the activation of STAT3, Akt and NF-κB Streptozotocin-induced diabetic [175]
mice
Propolis Enhance healing by increasing collagen production via TGF β1 and smad2, 3 signaling Streptozotocin-induced diabetic [79]
mice

10
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

Table 3
Marketed Products for rapid wound healing in diabetes.
Product Composed of company

Apligraf (Graftskin) Bovine collagen and living fibroblasts and keratinocytes Novartis, Switzerland
Dermagraft Cryopreserved human fibroblasts-derived dermal substitute Shireplc, USA
Becaplermin (Regranex) Platelet Derived Growth Factor-BB Smith & Nephew, Inc., USA
Bilayered living human skin equivalent Cultured keratinocytes on the fibroblast-populated collagen lattice
Angipars Melilotus officinalis Endocrinology and Metabolism Research Institute, Iran
Ampucare Azadirachta indica and Curcuma longa Venus Remedies Ltd., India
Fiblast Spray (Trafermin) Recombinant bovine bFGF Kaken Pharmaceutical Co.,Ltd.
MediHoney 80% active Leptospermum honey with colloidal alginate Derma Sciences, Inc., USA
rHuHSP90a-115 Topical protein drug
Woulgan® biogel (in clinical trial) Biotec Pharmacon’s soluble yeast beta-glucan (SBG) Biotec Pharmacon, Norway

Table 4
List of clinical trials studies on diabetic wound.
S. No. Study design Drugs/ Methods Result References

1. Randomized phase III Clinical Topical betulin gel Accelerates re-epithelialization of partial thickness wounds [176]
Trials
2. Randomized Clinical Trials Honey dressing In treated group, microbial clearance, and healing area were notably higher than control [177]
groups.
3. Randomized Clinical Trials Royal Jelly Healing area, healing rate and time not showing any change with placebo treated group. [178]
4. Randomized Clinical Trials Manuka Healing time of treated group was considerably lower than control group. And % of [179]
honey-impregnated dressing ulcers healed did not change significantly between groups
5. Randomized, Dragon's blood cream Dragon's blood cream significantly improves healing duration. [180]
Double-blind
6. Single-blinded randomized Extracorporeal Shock wave Noteworthy decrease in wound size and median time requisite for ulcer healing. [181]
controlled therapy
7. Single-arm clinical trial Autologous platelet-rich Wound area significantly decreased in treated group as compared to control. [182]
plasma gel
8. Randomized Controlled trial LeucoPatch system LeucoPatch treated group, 34% ulcers healed within 20 weeks as compared to 22% [183]
ulcers in the standard care group.
9. Randomized Controlled trial Negative pressure wound Reduces the granulation time of by 40% in diabetic wounds as compared to standard [184]
therapy wound dressing.

8. Conclusion UGC-BSR-7-341-2011 for JRF; UGC-MRP-41-748-2012 and DHR-ICMR


no. G.30011/4/2014-HR-2016 for financial support,
There has been exponential growth of research in the field of dia-
betic wound management over the past years. Impaired wound healing References
is common impediment of diabetes that has potentially devastating
consequences on suffering patients. Numerous factors contribute to [1] World Health Organization, Facts and Figures About Diabetes, Available at: (2014)
impaired healing in diabetes as suggested by various researches. http://www.who.int/mediacentre/factsheets/fs312/en/.2014.
[2] World Health Organization, Facts and Figures About Diabetes, Available at: (2013)
Significant developments have been made on various new therapeutic 2013 http://www.who.int/diabetes/facts/en/.
approaches and products in management of wound healing in diabetes. [3] V. Vijayakumar, S.K. Samal, S. Mohanty, S.K. Nayak, Recent advancements in
Approaches involving the growth factor, dual growth factors, various biopolymer and metal nanoparticle-based materials in diabetic wound healing
management, Biomacromolecules (2018), https://doi.org/10.1016/j.ijbiomac.
cytokines modulators, anti-inflammatory drugs, MMP inhibitors, an- 2018.10.120.
giogenesis stimulator, ECM stimulators, stem cells, and various natural [4] A. Mohandas, B.S. Anisha, K.P. Chennazhi, R. Jayakumar, Chitosan-hyaluronic
based products have been evaluated with limited achievement. Recent acid/VEGF loaded fibrin nanoparticles composite sponges for enhancing angio-
genesis in wounds, Colloids Surf.: B Biointerface 27 (2015) 105–113.
studies based on combinational approach have overpowered conven- [5] B.S. Anisha, R. Biswas, K.P. Chennazhi, R. Jayakumar, Chitosan-hyaluronic acid/
tional approaches. It presents a hope for the researchers to go through nano silver composite sponges for drug resistant bacteria infected diabetic
the new advancement in the designing of novel carrier along with un- wounds, Int. J. Biol. Macromol. 62 (2013) 310–320.
[6] A. Komarcevic, The modern approach to wound treatment, Med. Pregl. 53 (2000)
derstanding basic approach. Combination approaches can be an im-
363–368.
portant area for future research in management of compromised [7] A. Sharp, J. Clark, Diabetes and its impact on wound healing, Nurs. Stand. 25
wounds. Thus, contributing towards accelerated healing of diabetic (2011) 41–47.
wounds. Strong research is needed to recognize different agents that [8] D. Singh, M. Singh, S. Saraf, S. Saraf, Development of delivery cargoes for deb-
riding enzymes effective in wound healing, Trends Appl. Sci. Res. 6 (2011)
could act at different phase of wound healing in diabetes. Getting better 863–876.
clinical methodologies and system can be of assistance in recognizing [9] M.R. Singh, S. Saraf, A. Vyas, V. Jain, D. Singh, Innovative approaches in wound
the extent of healing. healing: trajectory and advances, Artif. Cell Nanomed. Biotechnol. 41 (2013)
202–212.
[10] A. Alavi, R.G. Sibbald, D. Mayer, L. Goodman, M. Botros, D.G. Armstrong, et al.,
Declaration of interest Dabetic foot ulcers part I. Pathophysiology and prevention, J. Am. Acad. Dermatol.
(2014) 1e1–1e16.
[11] H. Galkowska, U. Wojewodzka, W.L. Olszewski, Chemokines, cytokines, and
The authors report no declarations of interest. growth factors in keratinocytes and dermal endothelial cells in the margin of
chronic diabetic foot ulcers, Wound Repair Regen. 14 (2006) 558–565.
[12] K.V. Kavitha, S. Tiwari, V.B. Purandare, S. Khedkar, S.S. Bhosale,
Acknowledgements
A.G. Unnikrishnan, Choice of wound care in diabetic foot ulcer: a practical ap-
proach, World J. Diabetes 5 (2014) 546–556.
The authors are thankful to Director, University Institute of [13] S.B. Mallik, B.S. Jayashree, R.R. Shenoy, Epigenetic modulation of macrophage
Pharmacy, Pt. Ravishankar Shukla University Raipur, Chhattisgarh, polarization- perspectives in diabetic wounds, J. Diabetes Complicat. (2018),

11
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

https://doi.org/10.1016/j.jdiacomp.2018.01.015. e36974.
[14] K. Maruyama, et al., Decreased macrophage number and activation lead to re- [41] H.C. Chong, J.S.K. Chan, C.Q. Goh, N.V. Gounko, X. Wang, S. Foo, et al.,
duced lymphatic vessel formation and contribute to impaired diabetic wound Angiopoietin-like 4 stimulates STAT3-mediated iNOS expression and enhances
healing, Am. J. Pathol. 170 (2007) 1178–1191. angiogenesis to accelerate wound healing in diabetic mice, Mol. Ther. 22 (9)
[15] S. Okizaki, Y. Ito, K. Hosono, K. Oba, H. Ohkubo, H. Amano, et al., Suppressed (2014) 1593–1604.
recruitment of alternatively activated macrophages reduces TGF-β1 and impairs [42] A.Y. Peleg, T. Weerarathna, J.S. McCarthy, T.M. Davis, Common infections in
wound healing in streptozotocin induced diabetic mice, Biomed. Pharmacother. diabetes: pathogenesis, management and relationship to glycaemic control,
70 (2015) 317–325. Diabetes Metab. Res. Rev. 23 (2007) 13.
[16] M.A.M. Loots, S.B. Kenter, F.L. Au, W.J.M.Y. Galen, E. Middelkoop, J.D. Bos, [43] K. Singh, N.K. Agrawal, S.K. Gupta, G. Mohan, S. Chaturvedi, K. Singh, Decreased
J.R. Mekkes, Fibroblasts derived from chronic diabetic ulcers differ in their re- expression of heat shock proteins may lead to compromised wound healing in type
sponse to stimulation with EGF, IGF-I, bFGF and PDGF-AB compared to controls, 2 diabetes mellitus patients, J. Diabetes Complicat. 29 (2015) 578–588.
Eur. J. Cell Biol. 81 (1991) 153–160. [44] K. Singh, N.K. Agrawal, S.K. Gupta, G. Mohan, S. Chaturvedi, K. Singh, Genetic
[17] H. Brem, M. Tomic-Canic, Cellular and molecular basis of wound healing in dia- and epigenetic alterations in Toll like receptor 2 and wound healing impairment in
betes, J. Clin. Invest. 117 (2007) 1219–1222. type 2 diabetes patients, J. Diabetes Complicat. 29 (2015) 222–229.
[18] C. Schurmann, I. Goren, A. Linke, J. Pfeilschifter, S. Frank, Deregulated unfolded [45] K. Smith, A. Collier, E.M. Townsend, L.E. O’Donnell, A.M. Bal, J. Butcher, et al.,
protein response in chronic wounds of diabetic ob/ob mice: a potential connection One step closer to understanding the role of bacteria in diabetic foot ulcers:
to inflammatory and angiogenic disorders in diabetes-impaired wound healing, characterising the microbiome of ulcers, BMC Microbiol. 16 (2016) 54–66.
Biochem. Biophys. Res. Commun. 446 (2014) 195–200. [46] J. Moura, J. Rodrigues, M. Gonçalves, C. Amaral, M. Lima, E. Carvalho, Impaired
[19] J. Moura, E. Borsheim, E. Carvalho, The role of MicroRNAs in diabetic compli- T-cell differentiation in diabetic foot ulceration, Cell Mol. Immunol. (2016) 21.
cations-special emphasis on wound healing, Genes (Basel) 29 (2014) 926–956. [47] Y. Abiko, D. Selimovic, The mechanism of protracted wound healing on oral
[20] S. Biswas, et al., Hypoxia inducible microRNA 210 attenuates keratinocyte pro- mucosa in diabetes, Bosnian J. Basic Med. Sci. 10 (2010) 186–191.
liferation and impairs closure in a murine model of ischemic wounds, Proc. Natl. [48] Y. Nishikori, H. Okunishi, N. Shiota, The role of mast cells in cutaneous wound
Acad. Sci. U. S. A. 107 (2010) 6976–6981. healing in streptozotocin-induced diabetic mice, Arch. Dermatol. Res. 306 (2014)
[21] Y.C. Chan, S. Roy, S. Khanna, C.K. Sen, Downregulation of endothelial microRNA- 823–835.
200b supports cutaneous wound angiogenesis by desilencing GATA binding pro- [49] A. Tellechea, E.C. Leal, A. Kafanas, M.E. Auster, S. Kuchibhotla, Y. Ostrovsky,
tein 2 and vascular endothelial growth factor receptor 2, Arterioscler. Thromb. F. Tecilazich, D. Baltzis, et al., Mast Cells Regulate Wound Healing in Diabetes,
Vasc. Biol. 32 (2012) 1372–1382. Diabetes 65 (2016) 2006–2019.
[22] S. Bhattacharya, R. Aggarwal, V.P. Singh, S. Ramachandran, M. Datta, [50] D. Bevan, E. Gherardi, T.P. Fan, D. Edwards, R. Warn, Diverse and potent activities
Downregulation of miRNAs during delayed wound healing in diabetes: role of of HGF/SF in skin wound repair, J. Pathol. 203 (2004) 831–838.
dicer, Mol. Med. 2 (2015) 847–860. [51] G. Grieb, D. Simons, L. Eckert, M. Hemmrich, G. Steffens, J. Bernhagen, et al.,
[23] B. Icli, C.S. Nabzdyk, J.L. Hernandez, M. Cahill, M.E. Auster, et al., Regulation of Levels of macrophage migration inhibitory factor and glucocorticoids in chronic
impaired angiogenesis in diabetic dermal wound healing by microRNA-26a, J. wound patients and their potential interactions with impaired wound endothelial
Mol. Cell Cardiol. 91 (2016) 151–159. progenitor cell migration, Wound Repair Regen. 20 (2012) 707–714.
[24] Z. Li, S. Guo, F. Yao, Y. Zhang, T. Li, Increased ratio of serum matrix metallo- [52] H. Ebaid, B. Abdel-salam, I. Hassan, J. Al-Tamimi, A. Metwalli, I. Alhazza, Camel
proteinase-9 against TIMP-1 predicts poor wound healing in diabetic foot ulcers, J. milk peptide improves wound healing in diabetic rats by orchestrating the redox
Diabetes Complicat. 27 (2013) 380–382. status and immune response, Lipids Health Dis. 14 (2015) 132–142.
[25] L. Pradhan, X. Cai, S. Wu, N.D. Andersen, M. Martin, J. Malek, et al., Gene ex- [53] M. Luong, Y. Zhang, T. Chamberlain, T. Zhou, J.F. Wright, K. Dower, et al.,
pression of pro-inflammatory cytokines and neuropeptides in diabetic wound Stimulation of TLR4 by recombinant HSP70 requires structural integrity of the
healing, J. Surg. Res. 167 (2011) 336–342. HSP70 protein itself, J. Inflamm. 9 (2012) 11.
[26] C.J.N. Verkleij, J.J.T.H. Roelofs, S.R. Havik, J.C.M. Meijers, P.F. Marx, The role of [54] K.W. Park, S.H. Han, J.P. Hong, S. Han, D. Lee, B.S. Kim, J.H. Ahn, J.W. Lee,
thrombin-activatable fibrinolysis inhibitor in diabetic wound healing, Thromb. Topical epidermal growth factor spray for the treatment of chronic diabetic foot
Res. 126 (2010) 442–446. ulcers: a phase III multicenter, double-blind, randomized, placebo controlled trial,
[27] N. Nass, K. Vogel, B. Hofmann, P. Presek, R.E. Silber, A. Simm, Glycation of PDGF Diabetes Res. Clin. Pract. 142 (2018) 335–344.
results in decreased biological activity, Int. J. Biochem. Cell Biol. 2010 (42) (2010) [55] S. Barrientos, O. Stojadinovic, M.S. Golinko, H. Brem, M. TomicCanic, Growth
749–754. factors and cytokines in wound healing, Wound Repair Regen. 16 (2008) 585–601.
[28] M. Gooyit, Z. Peng, W.R. Wolter, H. Pi, D. Ding, D. Hesek, M. Lee, et al., A che- [56] G. Crovetti, G. Martinelli, M. Issi, M. Barone, M. Guizzardi, et al., Platelet gel for
mical biological strategy to facilitate diabetic wound healing, ACS Chem. Biol. 9 healing cutaneous chronic wounds, Transfus. Apher. Sci. 30 (2004) 145–151.
(2014) 105–110. [57] S. Frank, G. Hubner, G. Breier, M.T. Longaker, D.G. Greenhalgh, S. Wener,
[29] M. Brownlee, The patho-biology of diabetic complications: a unifying mechanism, Regulation of vascular endothelial growth factor expression in cultured kerati-
Diabetes 54 (2005) 1615–1625. nocytes. Implications for normal and impaired wound healing, J. Biol. Chem. 270
[30] S.K. Niture, A.K. Jaiswal, Inhibitor of Nrf2 (INrf2 or Keap1) protein degrades Bcl- (1995) 12607–12613.
xL via phosphoglycerate mutase 5 and controls cellular apoptosis, J. Biol. Chem. [58] R.L. Brown, M.P. Breeden, D.G. Greenhalgh, PDGF and TGFα act synergistically to
286 (2014) 44542–44556. improve wound healing in genetically diabetic mouse, J. Surg. Res. 56 (1994)
[31] M. Long, M. Rojo de la Vega, Q. Wen, M. Bharara, T. Jiang, R. Zhang, S. Zhou, 62–70.
et al., An essential role of NRF2 in diabetic wound healing, Diabetes 65 (2016) [59] M.S. Bitar, Z.N. Labbad, Transforming growth factor-β and insulin-like growth
780–793. factor I in relation to diabetes-induced impairment of wound healing, J. Surg. Res.
[32] S. Nakagomi, Y. Suzuki, K. Namikawa, S. Kiryu-Seo, H. Kiyama, Expression of 6 (1996) 113–119.
theactivating transcription factor 3 prevents c-Jun N-terminal kinase-induced [60] S. Werner, M. Breeden, G. Hubner, et al., Induction of keratinocyte growth factor
neuronal death by promoting heat shock protein 27 expression and Akt activation, expression is reduced and delayed during wound healing in the genetically dia-
J. Neurosci. 23 (2003) 5187–5196. betic mouse, J. Invest. Dermatol. 103 (1994) 469–473.
[33] G. Badr, W.N. Hozzein, H.M.S. Eldien, B.M. Badr, O. Garraud, Bee venom accel- [61] H.D. Beer, M.T. Longaker, S. Werner, Reduced expression of PDGF and PDGF re-
erates wound healing in diabetic mice by suppressing activating transcription ceptors during impaired wound healing, J. Invest. Dermatol. 109 (1997) 132–138.
factor-3 (ATF-3) and inducible nitric oxide synthase (iNOS)-mediated oxidative [62] N.L. Burstein, Growth factor effects on corneal wound healing, J. Ocular
stress and recruiting bone marrow derived endothelial progenitor cells, J. Cell. Pharmacol. Ther. 3 (1987) 263–277, https://doi.org/10.1089/jop.1987.3.263.
Physiol. 9999 (2016) 1–13. [63] R. Hosokawa, K. Kikuzaki, T. Kimoto, T. Matsuura, D. Chiba, M. Wadamoto, et al.,
[34] G. Badr, Camel whey protein enhances diabetic wound healing in a streptozotocin- Controlled local application of basic fibroblast growth factor (FGF-2) accelerates
induced diabetic mouse model: the critical role of β-Defensin-1, -2 and -3, Lipids the healing of GBR. An experimental study in beagle dogs, Clin. Oral Implants Res.
Health Dis. 12 (2013) 46–57. 11 (2000) 345–353.
[35] O. Dewald, P. Zymek, K. Winkelmann, A. Koerting, G. Ren, et al., CCL2/Monocyte [64] M.H. Parkar, L. Kuru, M. Giouzeli, I. Olsen, Expression of growth-factor receptors
chemoattractant Protein-1 regulates inflammatory responses critical to healing in normal and regenerating human periodontal cells, Arch. Oral Biol. 46 (2001)
myocardial infarcts, Circ. Res. 96 (2005) 881–889. 275–284.
[36] Y. Ishida, J.L. Gao, P.M. Murphy, Chemokine receptor CX3CR1 mediates skin [65] D.G. Greenhalgh, K.H. Sprugel, M.J. Murray, R. Ross, PDGF and FGF stimulate
wound healing by promoting macrophage and fibroblast accumulation and func- wound healing in the genetically diabetic mouse, Am. J. Pathol. 136 (1990)
tion, J. Immunol. 180 (2008) 569–579. 1235–1246.
[37] G. Badr, Supplementation with undenatured whey protein during diabetes mel- [66] D.L. Doxey, M.C. Ng, R.E. Dill, A.M. Iacopino, Platelet-derived growth factor levels
litus improves the healing and closure of diabetic wounds through the rescue of in wounds of diabetic rats, Life Sci. 57 (1995) 1111–1123.
functional long-lived wound macrophages, Cell. Physiol. Biochem. 29 (2012) [67] H. Li, X. Fu, L. Zhang, Q. Huang, Z. Wu, T. Sun, Research of PDGF-BB gel on the
571–582. wound healing of diabetic rats and its pharmacodynamics, J. Surg. Res. 145
[38] W. Clayton, A.Tom Elasy, A review of the patophysiology, classification, and (2008) 41–48.
treatment of foot ulcers in diabetic patients, Clin. Diabetes 27 (2009) 52–53. [68] R. Lobmann, C. Zemlin, M. Motzkau, K. Reshke, H. Lehnert, Expression of matrix
[39] H. Huang, W. Cui, W. Qiu, M. Zhu, R. Zhao, D. Zeng, et al., Impaired wound metalloproteinases and growth factors in diabetic foot wounds treated with a
healing results from the dysfunction of the Akt/mTOR pathway in diabetic rats, J. protease absorbent dressing, J. Diabetes Complicat. 20 (2006) 329–335.
Dermatol. Sci. (2015), https://doi.org/10.1016/j.jdermsci.2015.06.002. [69] R. Tsuboi, D.B. Rifkin, Recombinant basic fibroblast growth factor stimulates
[40] M.H.M. Lima, A.M. Caricilli, L.L. de Abreu, E.P. Araujo, F.F. Pelegrinelli, et al., wound healing in healing-impaired db/db mice, Int. J. Clin. Exp. Med. Res. 172
Topical insulin accelerates wound healing in diabetes by enhancing the AKT and (1990) 245–251.
ERK pathways: a double-blind placebo-controlled clinical trial, PLoS One 7 (2012) [70] L.G. Presta, H. Chen, S.J. O’Connor, V. Chisholm, Y.G. Meng, et al., Humanization

12
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

of an anti-vascular endothelial growth factor monoclonal antibody for the therapy release of basic fibroblast growth factor in a diabetic mouse model, J. Tissue Eng.
of solid tumors and other disorders, Cancer Res. 57 (1997) 4593–4599. Regen. Med. 8 (2014) 29–40.
[71] L.S. Angelo, R. Kurzrock, Vascular endothelial growth factor and its relationship to [100] P. Koria, H. Yagi, Y. Kitagawa, Z. Megeed, Y. Nahmias, R. Sheridan, et al., Self-
inflammatory mediators, Clin. Cancer Res. 13 (2007) 2825–2830. assembling elastin-like peptides growth factor chimeric nanoparticles for the
[72] K. Zhou, Y. Ma, M.S. Brogan, Chronic and non-healing wounds: the story of vas- treatment of chronic wounds, Proc. Natl. Acad. Sci. 108 (2011) 1034–1039.
cular endothelial growth factor, Med. Hypotheses 85 (2015) 399–404. [101] Y. Yang, T. Xia, F. Chen, W. Wei, C. Liu, S. He, et al., Electrospunfibers with
[73] J.T. Hardwicke, J. Hart, A. Bell, R. Duncan, D.W. Thomas, R. Moseley, The effect plasmid bFGFpolyplex loadings promote skin wound healing in diabetic rats, Mol.
of dextrin-rhEGF on the healing of full-thickness, excisional wounds in the (db/db) Pharm. 9 (2012) 48–58.
diabetic mouse, J. Control. Release 152 (2011) 411–417. [102] Z. Xie, C.B. Paras, H. Weng, P. Punnakitikashem, L.C. Su, et al., Dual growth factor
[74] D.L. Brown, C.D. Kane, S.D. Chernausek, et al., Differential expression and loca- releasing multi-functional nanofibers for wound healing, Acta Biomater. 9 (2013)
lization of insulin-like growth factors I and II in cutaneous wounds of diabetic and 9351–9359.
non-diabetic mice, Am. J. Pathol. 151 (1997) 715–724. [103] R. Judith, M. Nithya, C. Rose, A.B. Mandal, Application of a PDGF- containing
[75] B. Bruhn-Olszewska, A. Korzon-Burakowska, M. Gabig-Ciminska, et al., Molecular novel gel for cutaneous wound healing, Life Sci. 87 (2010) 1–8.
factors involved in the development of diabetic foot syndrome, Acta Biochim. Pol. [104] R.K. Chan, P.H. Liu, G. Pietramaggiori, S.I. Ibrahim, H.B. Hechtman, D.P. Orgill,
59 (2012) 507–513. Effect of recombinant platelet-derived growth factor (Regranex) on wound closure
[76] F.Y. Bhora, B.J. Dunkin, S. Batzri, H.M. Aly, B.L. Bass, A.N. Sidawy, et al., Effect of in genetically diabetic mice, J. Burn. Care Res. 27 (2006) 202–205.
growth factors on cell proliferation and epithelialization in human skin, J. Surg. [105] K. Mizuno, K. Yamamura, K. Yano, T. Osada, S. Saeki, N. Takimoto, et al., Effect of
Res. 59 (1995) 236–244. chitosan film containing basic fibroblast growth factor on wound healing in ge-
[77] A.B. Roberts, Transforming growth factor-β: activity and efficacy in animal models netically diabetic mice, J. Biomed. Mater. Res. 64 (2003) 177–181.
of wound healing, Wound Repair Regen. 3 (1995) 408–418. [106] D.G. Pyun, H.J. Choi, H.S. Yoon, T. Thambi, D.S. Lee, Polyurethane foam con-
[78] O. Stojadinovic, I. Pastar, K.A. Gordon, M. Tomic-Canic, Physiology and patho- taining rhEGF as a dressing material for healing diabetic wounds: synthesis,
physiology of wound healing in diabetes, in: A. Veves, J.M. Giurini, F.W. LoGerfo characterization, in vitro and in vivo studies, Colloids Surf. B: Biointerface 135
(Eds.), The Diabetic Foot. Humana Press Inc, New York, 2000, pp. 127–149. (2015) 699–706.
[79] W.N. Hozzein, G. Badr, A.A. Al Ghamdi, A. Sayed, N.S. Al-Waili, O. Garraud, [107] K.K. Chereddy, A. Lopes, S. Koussoroplis, V. Payen, C. Moia, et al., Combined
Topical application of Propolis Enhances cutaneous wound healing by promoting effects of PLGA and vascular endothelial growth factor promote the healing of
TGF-Beta/Smad-Mediated collagen production in a streptozotocin-induced type I non-diabetic and diabetic wounds, Nanomedicine (2015) 1–11, https://doi.org/
diabetic mouse model, Cell. Physiol. Biochem. 37 (2015) 940–954. 10.1016/j.nano.2015.07.006.
[80] E.B. Jude, Transforming growth factor-beta 1, 2, 3 and receptor type I and II in [108] T. Laing, R. Hanson, F. Chan, D. Bouchier-Hayes, Effect of pravastatin on ex-
diabetic foot ulcers, Diabetes Med. 19 (2002) 440–447. perimental diabetic wound healing, J. Surg. Res. 161 (2010) 336–340.
[81] H. Heublein, A. Bader, S. Giri, Preclinical and clinical evidence for stem cell [109] M. Bagheri, B.M. Jahromi, H. Mirkhani, Z. Solhjou, A. Noorafshan, et al.,
therapies as treatment for diabetic wounds, Drug Discov. Today 20 (2015) 6. Azelnidipine, a new calcium channel blocker, promotes skin wound healing in
[82] D.G. Armstrong, E.B. Jude, The role of matrix metalloproteinases in wound diabetic rats, J. Surg. Res. 169 (2011) e101–e107.
healing, J. Am. Podiatr. Med. Assoc. 92 (2002) 12–18. [110] Y. Lee, J.J. Chang, M.C. Yang, C.T. Chien, W.F. Lai, Acceleration of wound healing
[83] R. Lobmann, T.T. Pap, A. Ambrosch, K. Waldmann, W. Kfnig, H. Lehnert, in diabetic rats by layered hydrogel dressing, Carbohydr. Polym. 88 (2012)
Differential effects of PDGF-BB on matrix metalloproteases and cytokine release in 809–819.
fibroblasts of Type 2 diabetic patients and normal controls in vitro, J. Diabetes [111] U.F. Aly, Preparation and evaluation of novel topical gel preparations for wound
Comp. 20 (2006) 105–112. healing in diabetics, Int. J. Pharm. Pharm. Sci. 4 (2012) 76–77.
[84] S.V. McLennan, D. Min, D.K. Yue, Matrix metalloproteinases and their roles in [112] S. Hamed, Y. Ullmann, M. Masoud, E. Hellou, Z. Khamaysi, L. Teot, Topical ery-
poor wound healing in diabetes, Wound Pract. Res. 16 (2008) 116–121. thropoietin promotes wound repair in diabetic rats, J. Invest. Dermatol. 130
[85] S.J. Wall, D. Bevan, D.W. Thomas, G. Keith, K.G. Harding, et al., Differential ex- (2010) 87–94.
pression of matrix metalloproteinase during impaired wound healing of the dia- [113] S. Babaei, N. Bayat, M. Nouruzian, M. Bayat, Pentoxifylline improves cutaneous
betes mouse, J. Invest. Dermatol. 119 (91-) (2002) 98. wound healing in streptozotocin-induced diabetic rats, Eur. J. Pharmacol. 700
[86] R.G. Sibbald, K.Y. Woo, The biology of chronic foot ulcers in persons with DM, (2013) 165–172.
Diabetes/Metabolism Research & Review 24 (2008) S25–S30. [114] Z. Hou, C. Nie, Z. Si, Y. Ma, Deferoxamine enhances neovascularization and ac-
[87] J.P. Tardivo, F. Adami, J.A. Correa, M.A.S. Pinhal, M.S. Baptista, A clinical trial celerates wound healing in diabetic rats via the accumulation of hypoxia-inducible
testing the efficacy of PDT in preventing amputation in diabetic patients, factor-1α, Diabetes Res. Clin. Pract. (2013) 62–71.
Photodiagnosis Photodyn. Ther. 11 (3) (2014) 342–350. [115] M. Ram, V. Singh, S. Kumawat, D. Kumar, M.C. Lingaraju, et al., Deferoxamine
[88] M.R. Asadi, G. Torkaman, M. Hedayati, M.R. Mohajeri-Tehrani, M. Ahmadi, modulates cytokines and growth factors to accelerate cutaneous wound healing in
R.F. Gohardani, Angiogenic effects of low-intensity cathodal direct current on is- diabetic rats, Eur. J. Pharmacol. 764 (2015) 9–21.
chemic diabetic foot ulcers: a randomized controlled trial, Diabetes Res. Clin. [116] J.R. Scott, R.N. Tamura, P. Muangman, F.F. Isik, C. Xie, N.S. Gibran, Topical
Pract. 127 (2017) 147–155. substance P increases inflammatory cell density in genetically diabetic murine
[89] Z. Soleimani, F. Hashemdokht, F. Bahmani, M. Taghizadeh, M. Memarzadeh, wounds, Wound Repair Regen. 16 (2008) 529–533.
Z. Asem, Clinical and metabolic response to flaxseed oil omega-3 fatty acids [117] V. Kant, A. Gopal, D. Kumar, N.N. Pathak, et al., Curcumin-induced angiogenesis
supplementation in patients with diabetic foot ulcer: a randomized, doubleblind, hastens wound healing in diabetic rats, J. Surg. Res. 193 (2015) 978–988.
placebo-controlled trial, J. Diabetes Complicat. 31 (9) (2017) 1394–1400. [118] E.C. Leal, E. Carvalho, A. Tellechea, A. Kafanas, F. Tecilazich, C. Kearney, et al.,
[90] Y. Yang, T. Xia, W. Zhi, L. Wei, J. Weng, C. Zhang, et al., Promotion of skin re- Substance P promotes wound healing in diabetes by modulating inflammation and
generation in diabetic rats by electrospun core-sheath fibers loaded with basic macrophage phenotype, Am. J. Pathol. 185 (2015) 1638–1648.
fibroblast growth factor, Biomaterials 32 (2011) 4243–4254. [119] B. Romana-Souza, A.P. Nascimento, A. Monte-Alto-Costa, Propranolol improves
[91] M.W. Tsang, W.K.R. Wong, C.S. Hung, K.M. Lai, W. Tang, et al., Human epidermal cutaneous wound healing in streptozotocin-induced diabetic rats, Eur. J.
growth factor enhances healing of diabetic foot ulcers, Diabetes Care 26 (2003) Pharmacol. 6 (11) (2009) 77–84.
1856–1862. [120] C.H. Lee, S.H. Chang, W.J. Chen, K.C. Hung, S.J. Liu, et al., Augmentation of
[92] X. Dong, J. Xu, W. Wang, H. Luo, X. Liang, L. Zhang, et al., Repair effect of diabetic diabetic wound healing and enhancement of collagen content using nano-fibrous
ulcers with recombinant human epidermal growth factor loaded by sustained-re- glucophage-loaded collagen/PLGA scaffold membranes, J. Colloid Interface Sci.
lease microspheres, Sci. China Life Sci. 51 (2008) 1039–1044. 439 (2015) 88–97.
[93] N. Morimoto, K. Yoshimura, M. Niimi, T. Ito, H. Tada, S. Teramukai, et al., An [121] P. Kaur, A.K. Sharma, D. Nag, A. Das, S. Datta, A. Ganguli, V. Goel, S. Rajput,
exploratory clinical trial for combination wound therapy with a novel medical G. Chakrabarti, B. Basu, D. Choudhury, Novel nano-insulin formulation modulates
matrix and fibroblast growth factor in patients with chronic skin ulcers: a study cytokine secretion and remodeling to accelerate diabetic wound healing,
protocol, Am. J. Transl. Res. 4 (2012) 52–59. Nanomedicine (2018), https://doi.org/10.1016/j.nano.2018.08.013.
[94] B. Cheng, H.W. Liu, X.B. Fu, T.Z. Sun, Z.Y. Sheng, Recombinant human platelet- [122] X. Wang, M.K. Sng, S. Foo, H.C. Chong, M.B.Y. Tang, et al., Early controlled release
derived growth factor enhanced dermal wound healing by a pathway involving of peroxisome proliferator-activated receptor β/δ agonist GW501516 improves
ERK and c-fos in diabetic rats, J. Dermatol. Sci. 45 (2007) 193–201. diabetic wound healing through redox modulation of wound microenvironment, J.
[95] X. Yan, B. Chen, Y. Lin, Y. Li, Z. Xiao, et al., Acceleration of diabetic wound healing Control. Release 197 (2015) 138–147.
by collagen-binding vascular endothelial growth factor in diabetic rat model, [123] A.J. Whittam, Z.N. Maan, D. Duscher, J.A. Barrera, M.S. Hu, L.H. Fischer, et al.,
Diabetes Res. Clin. Pract. 90 (2010) 66–72. Small molecule inhibition of dipeptidyl Peptidase-4 enhances bone marrow pro-
[96] W. Wang, S. Lin, Y. Xiao, Y. Huang, Y. Tan, L. Cai, X. Li, Acceleration of diabetic genitor cell function and angiogenesis in diabetic wounds, Transl. Res. (2018),
wound healing with chitosan crosslinked collagen sponge containing recombinant https://doi.org/10.1016/j.trsl.2018.10.006.
human acidic fibroblast growth factor in healing-impaired STZ diabetic rats, Life [124] F. Lv, J. Wang, P. Xu, Y. Han, H. Ma, H. Xu, S. Chen, J. Chang, Q. Ke, M. Liu, Z. Yi,
Sci. 82 (2008) 190–204. C. Wu, A conducive bioceramic/polymer composite biomaterial for diabetic
[97] J.S. Choi, K.W. Leong, H.S. Yoo, In vivo wound healing of diabetic ulcers using wound healing, Acta Biomater. (2017), https://doi.org/10.1016/j.actbio.2017.07.
electrospun nanofibers immobilized with human epidermal growth factor (EGF), 020.
Biomaterials 29 (2008) 587–596. [125] J.G. Leu, M.H. Chiang, C.Y. Chen, J.T. Lin, H.M. Chen, Y.L. Chen, Y.L. Liang,
[98] Y. Matsumoto, Y. Kuroyanagi, Development of a wound dressing composed of Adenine accelerated the diabetic wound healing by PPAR delta and angiogenic
hyaluronic acid sponge containing arginine and epidermal growth factor, J. regulation, Eur. J. Pharmacol. (2017), https://doi.org/10.1016/j.ejphar.2017.11.
Biomater. Sci. Polym. Ed. 21 (2010) 715–726. 027.
[99] N. Kanda, N. Morimoto, A.A. Ayvazyan, S. Takemoto, K. Kawai, Y. Nakamura, [126] N.H. Wael, G. Badr, B.M. Badre, A. Allam, et al., Bee venom improves diabetic
et al., Evaluation of a novel collagen-gelatin scaffold for achieving the sustained wound healing by protecting functional macrophages from apoptosis and

13
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

enhancing Nrf2, Ang-1 and Tie-2 signaling, Mol. Immunol. 103 (2018) 322–335. [152] M.A. Abu-Al-Basal, Healing potential of Rosmarinus officinalis L. On full-thickness
[127] U. Bulbake, S. Jain, N. Kumar, A. Mittal, Curcumin loaded biomimetic composite excision cutaneous wounds in alloxan-induced-diabetic BALB/c mice, J.
graft for faster regeneration of skin in diabetic wounds, J. Drug Deliv. Sci. Technol. Ethnopharmacol. 131 (2010) 443–450.
(2018), https://doi.org/10.1016/j.jddst.2018.06.016. [153] A. Singha, S. Bajpaia, N. Singha, V. Kumara, J.K. Goura, et al., Wound healing
[128] T. Kawanabe, T. Kawakami, Y. Yatomi, S. Shimada, Y. Soma, Sphingosine 1- activity of standardized extract of Curculigo orchioides in streptozotocin–induced
phosphate accelerates wound healing in diabetic mice, J. Cosmet. Dermatol. Sci. diabetic mice, Asian Pac. J. Trop. Dis. 4 (2014) S48–S53.
Appl. 48 (2007) 53–60. [154] B. Larijani, R. Heshmat, A. Bahrami, H. Delshad, et al., Effect of intravenous
[129] K. Kawai, A. Kageyama, T. Tsumano, S. Nishimoto, K. Fukuda, et al., Effects of Semelil (ANGIPARSTM) on diabetic foot ulcers healing: a multicenter clinical trial,
adiponectin on growth and differentiation of human keratinocytes-Implication of Daru J. Pharm. Sci. 16 (2008) 35–40.
impaired wound healing in diabetes, Biochem. Biophys. Res. Commun. 374 (2008) [155] T.W. Lau, F.F.Y. Lam, K.M. Lau, Y.W. Chan, K.M. Lee, et al., Pharmacological
269–273. investigation on the wound healing effects of Radix rehmanniae in an animal
[130] L.I.F. Moura, A.M.A. Dias, E.C. Leal, L. Carvalho, H.C. De Sousa, E. Carvalho, model of diabetic foot ulcer, J. Ethnopharmacol. 123 (2009) 155–162.
Chitosan-based dressings loaded with Neurotensin—an efficient strategy to im- [156] J.G. Merrel, S.W. McLaughlin, L. Tie, C.T. Laurencin, A.F. Chen, L.S. Nair,
prove early diabetic wound healing, Acta Biomater. 2 (2014) 843–857. Curcumin loaded poly(e-caprolactone) nanofibres: diabetic wound dressing with
[131] L.I.F. Moura, A.M.A. Dias, E. Suesca, S. Casadiegos, E.C. Leal, et al., Neurotensin- anti-oxidant and anti-inflammatory properties, Clin. Exp. Pharmacol. Physiol.
loaded collagen dressings reduce inflammation and improve wound healing in Suppl. 36 (2009) 1149–1156.
diabetic mice, Biochim. Biophys. Acta 1842 (2014) 32–43. [157] V. Kant, A. Gopal, N.N. Pathak, P. Kumar, et al., Antioxidant and anti-infl-
[132] M. Galeano, F. Polito, A. Bitto, N. Irrera, G.M. Campo, et al., Systemic adminis- ammatory potential of curcumin accelerated the cutaneous wound healing in
tration of high-molecular weight hyaluronan stimulates wound healing in ge- streptozotocin-induced diabetic rats, Int. Immunopharmacol. 20 (2014) 322–330.
netically diabetic mice, Biochim. Biophys. Acta 1812 (2011) 752–759. [158] V. Kant, D. Kumar, D. Kumar, R. Prasad, A. Gopal, P. Kumar, et al., Topical ap-
[133] S.L. Hansen, C.A. Myers, A. Charboneau, D.M. Young, N. Boudreau, HoxD3 ac- plication of substance P promotes wound healing in streptozotocin-induced dia-
celerates wound healing in diabetic mice, Am. J. Pathol. 163 (2003) 2421–2431. betic rats, Cytokine 73 (2015) 144–155.
[134] A. Bazrafshan, M. Owji, M. Yazdani, M. Varedi, Activation of mitosis and angio- [159] S. Lodhi, A.K. Singhai, Wound healing effect of flavonoid rich fraction and luteolin
genesis in diabetes-impaired wound healing by processed human amniotic fluid, J. isolated from Martynia annua Linn. On streptozotocin induced diabetic rats, Asian
Surg. Res. 188 (2014) 545–552. Pac. J. Trop. Med. (2013) 253–259.
[135] M.K. Maharlooei, M. Bagheri, Z. Solhjou, B.M. Jahromi, M. Akrami, et al., Adipose [160] L. Tie, Y. An, J. Han, Y. Xiao, Y. Xiaokaiti, S. Fan, S. Liu, A.F. Chen, X. Li, Genistein
tissue derived mesenchymal stem cell (AD-MSC) promotes skin wound healing in accelerates refractory wound healing by suppressing superoxide and FoxO1/iNOS
diabetic rats, Diabetes Res. Clin. Pract. 93 (2001) 228–234. pathway in type 1 diabetes, J. Nutr. Biochem. 24 (2013) 88–96.
[136] P. Velander, C. Theopold, O. Bleiziffer, et al., Cell suspensions of autologous [161] N. Fujita, I. Sakaguchi, H. Kobayashi, N. Ikeda, Y. Kato, et al., An extract of the
keratinocytes or autologous fibroblasts accelerate the healing of full thickness skin root of Lithospermunerythrorhison accelerates wound healing in diabetic mice,
wounds in a diabetic porcine wound healing model, J. Surg. Res. 157 (2009) Biol. Pharm. Bull. 26 (2003) 329–335.
14–20. [162] J.C.W. Tam, C.H. Ko, K.M. Lau, M.H. To, H.F. Kwok, et al., A Chinese 2-herb
[137] S. Kazemi-Darabadi, F. Sarrafzadeh-Rezaei, A.A. Farshid, B. Dalir-Naghadeh, formula (NF3) promotes hindlimb ischemia-induced neovascularization and
Allogenous skin fibroblast transplantation enhances excisional wound healing wound healing of diabetic rats, J. Diabetes Complicat. 28 (2014) 436–447.
following alloxan diabetes in sheep, a randomized controlled trial, Int. J. Surg. 12 [163] J.C.W. Tam, K.M. Lau, C.L. Liu, M.H. To, H.F. Kwok, et al., The in vivo and in vitro
(2014) 751–756. diabetic wound healing effects of a 2-herb formula and its mechanisms of action, J.
[138] V. Falanga, S. Iwamoto, M. Chartier, T. Yufit, J. Butmarc, N. Kouttab, et al., Ethnopharmacol. 134 (2011) 831–838.
Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a [164] T. Ponrasu, L. Suguna, Efficacy of Annona squamosa on wound healing in strep-
fibrin spray accelerate healing in murine and human cutaneous wounds, Tissue tozotocin-induced diabetic rats, Int. Wound J. 9 (2012) 613–623.
Eng. 13 (2007) 1299–1312. [165] J. Jacobi, J.J. Jang, U. Sundram, H. Dayoub, L.F. Fajardo, J.P. Cooke, Nicotine
[139] Y.R. Kuo, C.T. Wang, J.T. Cheng, et al., Bone marrow-derived mesenchymal stem accelerates angiogenesis and wound healing in genetically diabetic mice, Am. J.
cells enhanced diabetic wound healing through recruitment of tissue regeneration Pathol. 161 (2002) 97–104.
in a rat model of streptozotocin-induced diabetes, Plast. Reconstr. Surg. 128 [166] M. Zuber, V. Rajesh, K. Anusha, C.R. Reddy, A. Tirupathi, Wound healing activity
(2011) 872–880. of ethanolic extract of Allium sativum on alloxan induced diabetic rats, Int. J. Sci.
[140] C. Gu, S. Huang, D. Gao, Y. Wu, J. Li, et al., Angiogenic effect of mesenchymal Invent. Today 2 (2013) 40–57.
stem cells as a therapeutic target for enhancing diabetic wound healing, Int. J. [167] A.D. Kandhare, P. Ghosh, S.L. Bodhankar, Naringin, a flavanone glycoside, pro-
Lower Extrem. Wounds 13 (2014) 88–93. motes angiogenesis and inhibits endothelial apoptosis through modulation of
[141] T.G. Nithya, Efficacy of mesenchymal stem cell incorporation in different com- inflammatory and growth factor expression in diabetic foot ulcer in rats, Chem.-
posite electrospun collagen nanofibers for diabetic wound healing, Asian J. Pharm. Biol. Interact. 219 (2014) 101–112.
Clin. Res. 6 (2013) 149–152. [168] P. Inpanya, A. Faikrua, A. Ounaroon, A. Sittichokechaiwut, J. Viyoch, Effects of
[142] K.B. Lee, J. Choi, S.B. Cho, J.Y. Chung, E.S. Moon, et al., Topical embryonic stem the blended fibroin/aloe gel film on wound healing in streptozotocin-induced
cells enhance wound healing in diabetic rats, J. Orthop. Res. 29 (2011) diabetic rats, Biomed. Mater. 7 (2012) 035008.
1554–1562. [169] A.H. Kwon, Z. Qiu, M. Hashimoto, K. Yamamoto, T. Kimura, Effects of medicinal
[143] S.W. Kim, H.Z. Zhang, L. Guo, J.M. Kim, M.H. Kim, Amniotic mesenchymal stem mushroom (Sparassis crispa) on wound healing in streptozotocin-induced diabetic
cells enhance wound healing in diabetic NOD/SCID mice through high angiogenic rats, Am. J. Surg. 197 (2009) 503–509.
and engraftment capabilities, PLoS One 7 (2012) e41105. [170] P. Molan, Honey Based Wound Dressings, USRE42755 E (2011).
[144] P. Kong, X. Xie, F. Li, Y. Liu, Y. Lu, Placenta mesenchymal stem cell accelerates [171] H. Mohamed, B.E.I. Lenjawi, M.A. Salma, S. Abdi, Honey based therapy for the
wound healing by enhancing angiogenesis in diabetic Goto-Kakizaki (GK) rats, management of a recalcitrant diabetic foot ulcer, J. Tissue Viability 23 (2014)
Biochem. Biophys. Res. Commun. 438 (2013) 410–419. 29–33.
[145] E.H. Javazon, S.G. Keswani, A.T. Badillo, T.M. Crombleholme, et al., Enhanced [172] Y. Yang, F. Wang, D. Yin, Z. Fang, L. Huang, Astragulus polysaccharide-loaded
epithelial gap closure and increased angiogenesis in wounds of diabetic mice fibrous mats promote the restoration of microcirculation in/around skin wounds
treated with adult murine bone marrow stromal progenitor cells, Wound Repair to accelerate wound healing in a diabetic rat model, Colloid Surf. B Biointerfaces
Regen. 15 (2007) 350–359. (2015), https://doi.org/10.1016/j.colsurfb.2015.09.006.
[146] M. Nambu, S. Kishimoto, S. Nakamura, H. Mizuno, S. Yanagibayashi, et al., [173] X. Zhang, Z. Ma, Y. Wang, Y. Li, B. Sun, X. Guo, C. Pan, L. Chen, The four-herb
Accelerated wound healing in healing-impaired db/db mice by autologous adipose chinese medicine formula Tuo-Li-Xiao-Du-San accelerates cutaneous wound
tissue-derived stromal cells combined with atelocollagen matrix, Ann. Plast. Surg. healing in streptozotocin-induced diabetic rats through reducing inflammation
62 (2009) 317–321. and increasing angiogenesis, J. Diabetes Res. 5 (2016) 639129, 11.
[147] D. Ma, J.E.H. Kua, W.K. Lim, S.T. Lee, A.W.C. Chua, In vitro characterization of [174] R.R. Ahmed, A. Mahmoud, O.M. Ahmed, A. Metwalli, H. Ebaid, Up-regulation of
human hair follicle dermal sheath mesenchymal stromal cells and their potential Hsp72 and keratin16 mediates wound healing in streptozotocin diabetic rats, Biol.
in enhancing diabetic wound healing, Cytotherapy 17 (2015) 1036–1051. Res. 48 (2015) 54–66.
[148] D.C.S. Pedroso, A. Tellechea, L. Moura, I. Fidalgo-Carvalh, J. Duarte, et al., [175] G. Badr, B.M. Badr, M.H. Mahmoud, M. Mohany, D.M. Rabah, O. Garraud,
Improved survival, vascular differentiation and wound healing potential of stem Treatment of diabetic mice with undenatured whey protein accelerates the wound
cells Co-cultured with endothelial cells, PLoS One 6 (2011) e16114. healing process by enhancing the expression of MIP-1α, MIP-2, KC, CX3CL1 and
[149] Q. Xu, A. Sigen, Y. Gao, L. Guo, J. Creagh-Flynn, D. Zhou, U. Greiser, Y. Dong, TGF-β in wounded tissue, BMC Immunol. 13 (2012) 32–41.
F. Wang, H. Tai, W. Liu, W. Wang, W. Wang, A hybrid injectable hydrogel from [176] J.P. Barret, F. Podmelle, B. Lipový, H.O. Rennekampff, H. Schumann,
hyperbranched PEG macromer as a stem cell delivery and retention platform for A. Schwieger-Briel, et al., Accelerated re-epithelialization of partial-thickness skin
diabetic wound healing, Acta Biomater. (2018), https://doi.org/10.1016/j.actbio. wounds by a topical betulin gel: results of a randomized phase III clinical trials
2018.05.039. program, Burns 4 (2017) 1284–1294.
[150] R.F. Irons, K.W. Cahill, D.A. Rattigan, J.H. Marcotte, M.W. Fromer, S. Shaohua [177] C.L. Guo, X.Y. Fu, Research on effect evaluation of local treatment of patients with
Chang, et al., Acceleration of diabetic wound healing with adipose-derived stem diabetic foot ulcers using honey dressing, Med. J. West China 7 (2013) 977–980.
cells, endothelial-differentiated stem cells, and topical conditioned medium [178] M. Siavash, S. Shokri, S. Haghighi, M.A. Shahtalebi, Z. Farajzadehgan, The efficacy
therapy in a swine model, J. Vasc. Surg. (2018), https://doi.org/10.1016/j.jvs. of topical royal jelly on healing of diabetic foot ulcers: a double-blind placebo-
2018.01.065. controlled clinical trial, Int. Wound J. 12 (2015) 137–142.
[151] K.C. Moon, J.S. Lee, S.K. Han, H.W. Lee, E.S. Dhong, Effects of human umbilical [179] A.V. Kamaratos, K.N. Tzirogiannis, S.A. Iraklianou, G.I. Panoutsopoulos,
cord blood–derived mesenchymal stromal cells and dermal fibroblasts on diabetic I.E. Kanellos, A.I. Melidonis, Manuka honey-impregnated dressings in the treat-
wound healing, Cytotherapy (2017), https://doi.org/10.1016/j.jcyt.2017.03.074. ment of neuropathic diabetic foot ulcers, Int. Wound J. 11 (2014) 259–263.

14
S. Patel, et al. Biomedicine & Pharmacotherapy 112 (2019) 108615

[180] F. Namjoyan, F. Kiashi, Z.B. Moosavi, F. Saffari, B.S. Makhmalzadeh, Efficacy of 10.020.
Dragon’s blood cream onwound healing: a randomized, double-blind, placebo [183] F. Game, W. Jeffcoate, L. Tarnow, J.L. Jacobsen, D.J. Whitham, E.F. Harrison,
controlled clinical trial, J. Tradit. Complement. Med. (2015) 1–4. S.J. Ellender, D. Fitzsimmons, M. Löndahl, LeucoPatch system for the management
[181] M.T.A. Omar, A. Alghadir, K.K. Al-Wahhabi, A.B. Al-Askar, Efficacy of shock wave of hard-to-heal diabetic foot ulcers in the UK, Denmark, and Sweden: an observer-
therapy on chronic diabetic foot ulcer: a single-blinded randomized controlled masked, randomised controlled trial, Lancet Diabetes Endocrinol. S2213-8587
clinical trial, Diabetes Res. Clin. Pract. (2014), https://doi.org/10.1016/j.diabres. (18) (2018) 30240–30247, https://doi.org/10.1016/.
2014.09.024. [184] G. Sepulveda, M. Espindola, M. Maureira, E. Sepúlveda, J.I. Fernández, C. Oliva,
[182] M.H. Mohammadi, B. Molavi, S. Mohammadi, M. Nikbakht, A.M. Mohammadi, A. Sanhueza, M. Vial, C. Manterola, Negative-pressure wound therapy versus
S. Mostafaei, A.H. Norooznezhad, A.G. Abdegah, A. Ghavamzadeh, Evaluation of standard wound dressing in the treatment of diabetic foot amputation: a rando-
wound healing in diabetic foot ulcer using platelet-rich plasma gel: a single-arm mised controlled trial, Cir. Esp. 86 (3) (2009) 171–177.
clinical trial, Transfus. Apher. Sci. (2016), https://doi.org/10.1016/j.transci.2016.

15

You might also like